1
|
Walker LA, Bagonza V, Bobb B, Sullivan DJ. Modified dosing schedule efficacy of fosmidomycin and clindamycin against murine malaria Plasmodium berghei. Int J Parasitol Drugs Drug Resist 2025; 27:100577. [PMID: 39746289 PMCID: PMC11754493 DOI: 10.1016/j.ijpddr.2024.100577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Fosmidomycin and clindamycin target the Plasmodium apicoplast. Combination clinical trials have produced mixed results with the primary problem being the recrudescent infection frequency by day 28. Given that antibiotic efficacy against bacterial infections often depends on the constant drug presence over several days, we hypothesized that the antimalarial blood or liver stage efficacy of fosmidomycin and clindamycin could be improved by implementing a more frequent dosing schedule. A blood stage murine malaria P. berghei GFP-luciferase low and high parasitemia model was implemented to follow pharmacodynamics and cure for modified dose, schedule and duration of individual and combination fosmidomycin and clindamycin. P. berghei sporozoites were used to investigate fosmidomycin during the 48 h murine liver stage. Here we observed that the same total dose of fosmidomycin and clindamycin, alone and in combination, are more efficacious when scheduled in smaller, more frequent doses. Fosmidomycin added measurably small additional killing in combination with clindamycin. Despite dosing every 6 h during liver stages, fosmidomycin was inhibitory, but noncurative even with addition of atorvastatin to decrease hepatocyte production of mevalonate. We have also demonstrated in vitro efficacy of fosmidomycin and clindamycin against P. falciparum C580Y with IC50s similar to those for drug sensitive P. falciparum. The dosing schedule of quinoline and artemisinin partner drugs fosmidomycin or clindamycin targeting the apicoplast should maximize time above minimum inhibitory concentration.
Collapse
Affiliation(s)
- Leah A Walker
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, 21210, USA
| | - Vision Bagonza
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, 21210, USA
| | - Bryce Bobb
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, 21210, USA
| | - David J Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD, 21210, USA.
| |
Collapse
|
2
|
Theuretzbacher U, Jumde RP, Hennessy A, Cohn J, Piddock LJV. Global health perspectives on antibacterial drug discovery and the preclinical pipeline. Nat Rev Microbiol 2025:10.1038/s41579-025-01167-w. [PMID: 40148602 DOI: 10.1038/s41579-025-01167-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
Antibacterial resistance is a global challenge that requires a coordinated international response. The current clinical pipeline largely consists of derivatives of established antibiotic classes, whereas the discovery and preclinical pipeline is diverse and innovative including new direct-acting agents with no cross-resistance with existing antibiotics. These novel compounds target pathways such as lipoprotein synthesis, lipopolysaccharide biosynthesis and transport, outer membrane assembly, peptidoglycan biosynthesis, fatty acid biosynthesis and isoprenoid biosynthesis. If these agents can be developed into safe, effective and affordable drugs, they could address a broad range of infections worldwide, benefiting large patient populations without geographical limitations. However, strategies such as indirect-acting or pathogen-specific treatments are likely to benefit small patient groups, primarily in high-income countries that have advanced health-care systems and diagnostic infrastructure. Although encouraging, the discovery and preclinical pipeline remains insufficiently robust to offset the high attrition rates typical of early-stage drug innovation and to meet global health needs.
Collapse
Affiliation(s)
| | - Ravindra P Jumde
- Global Antibiotic Research and Development Partnership, Geneva, Switzerland
| | - Alan Hennessy
- Global Antibiotic Research and Development Partnership, Geneva, Switzerland
| | - Jennifer Cohn
- Global Antibiotic Research and Development Partnership, Geneva, Switzerland
| | - Laura J V Piddock
- Global Antibiotic Research and Development Partnership, Geneva, Switzerland.
| |
Collapse
|
3
|
Zhou Y, Wang J, Sun Y, Cheng Y, Wu W. Non-Hydroxamate Inhibitors of IspC Enzyme in the MEP Pathway: Structural Insights and Drug Development Potential. Chem Biol Drug Des 2025; 105:e70086. [PMID: 40099748 DOI: 10.1111/cbdd.70086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
1-Deoxy-D-xylulose-5-phosphate reductoisomerase (IspC) is a key enzyme in the MEP pathway, essential for many bacteria, human pathogens, and plants, thus being an attractive drug target. Fosmidomycin, a potent IspC inhibitor with hydroxamate metal-binding pharmacophores (MBPs), has entered clinical trials for malaria but is hampered by pharmacokinetic and toxicity issues of the hydroxamate fragment. This has led to increased interest in non-hydroxamate inhibitors. This review focuses on the crystal structure and active-site binding mode of IspC, and the structural types, inhibitory activities, and structure-activity relationships of non-hydroxamate IspC inhibitors. Early attempts to design such inhibitors involved direct removal or replacement of the hydroxamate MBPs, with varying results. Lipophilic inhibitors, bisubstrate inhibitors, and those developed for herbicidal applications have shown promise. However, challenges remain due to the sensitivity of the enzyme active site to ligand interactions. Future research could draw from other metalloenzyme studies to develop novel and efficient non-hydroxamate IspC inhibitors.
Collapse
Affiliation(s)
- Yaqing Zhou
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Jili Wang
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Yong Sun
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Yarui Cheng
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Wenhai Wu
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| |
Collapse
|
4
|
Knak T, Takada S, Illarionov B, Krisilia V, Pessanha de Carvalho L, Lungerich B, Sakamoto Y, Höfmann S, Bacher A, Kalscheuer R, Held J, Fischer M, Tanaka N, Kurz T. Expanding the Chemical Space of Reverse Fosmidomycin Analogs. ACS Med Chem Lett 2025; 16:136-143. [PMID: 39811140 PMCID: PMC11726376 DOI: 10.1021/acsmedchemlett.4c00501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Multidrug-resistant pathogens pose a major threat to human health, necessitating the identification of new drug targets and lead compounds that are not susceptible to cross-resistance. This study demonstrates that novel reverse thia analogs of the phosphonohydroxamic acid antibiotic fosmidomycin inhibit 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR), an essential enzyme for Plasmodium falciparum, Escherichia coli, and Mycobacterium tuberculosis that is absent in humans. Some novel analogs with large α-phenyl substituents exhibited strong inhibition across these three DXR orthologues, surpassing the inhibitory activity of fosmidomycin. Despite nanomolar target inhibition, the new DXR inhibitors demonstrated mainly weak or no in vitro growth inhibition of the pathogens. Crystallographic studies revealed that compounds 12a and 12b induce an open PfDXR conformation and that the enzyme selectively binds the S-enantiomers. The study underscores the difficulties of achieving potent cellular activity despite strong DXR inhibition and emphasizes the need for novel structural optimization strategies and comprehensive pharmacokinetic studies.
Collapse
Affiliation(s)
- Talea Knak
- Institute
of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Sana Takada
- School
of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641, Japan
| | - Boris Illarionov
- Hamburg
School of Food Science, Universität
Hamburg, Grindelallee
117, 20146 Hamburg, Germany
| | - Violetta Krisilia
- Institute
of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr.
1, 40225 Düsseldorf, Germany
| | - Lais Pessanha de Carvalho
- Institut
für Tropenmedizin, Eberhard Karls
Universität Tübingen, Wilhelmstr. 27, 72074 Tübingen, Germany
| | - Beate Lungerich
- Institute
of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Yasumitsu Sakamoto
- School
of
Pharmacy, Iwate Medical University, Yahaba, Iwate 028-3694, Japan
| | - Stefan Höfmann
- Institute
of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Adelbert Bacher
- Hamburg
School of Food Science, Universität
Hamburg, Grindelallee
117, 20146 Hamburg, Germany
- TUM
School of Natural Sciences, Technical University
of Munich, Boltzmannstr.
10, 85748 Garching, Germany
| | - Rainer Kalscheuer
- Institute
of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr.
1, 40225 Düsseldorf, Germany
| | - Jana Held
- Institut
für Tropenmedizin, Eberhard Karls
Universität Tübingen, Wilhelmstr. 27, 72074 Tübingen, Germany
- German
Center for Infection Research (DZIF),
partner site Tübingen, 72074 Tübingen, Germany
| | - Markus Fischer
- Hamburg
School of Food Science, Universität
Hamburg, Grindelallee
117, 20146 Hamburg, Germany
| | - Nobutada Tanaka
- School
of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641, Japan
| | - Thomas Kurz
- Institute
of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics
and Natural Sciences, Heinrich Heine University
Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| |
Collapse
|
5
|
Takada S, Abdullaziz MA, Höfmann S, Knak T, Ozawa SI, Sakamoto Y, Kurz T, Tanaka N. The Diverse Binding Modes Explain the Nanomolar Levels of Inhibitory Activities Against 1-Deoxy-d-Xylulose 5-Phosphate Reductoisomerase from Plasmodium falciparum Exhibited by Reverse Hydroxamate Analogs of Fosmidomycin with Varying N-Substituents. Molecules 2024; 30:72. [PMID: 39795129 PMCID: PMC11721986 DOI: 10.3390/molecules30010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
It is established that reverse hydroxamate analogs of fosmidomycin inhibit the growth of Plasmodium falciparum by inhibiting 1-deoxy-d-xylulose 5-phosphate reductoisomerase (DXR), the second enzyme of the non-mevalonate pathway, which is absent in humans. Recent biochemical studies have demonstrated that novel reverse fosmidomycin analogs with phenylalkyl substituents at the hydroxamate nitrogen exhibit inhibitory activities against PfDXR at the nanomolar level. Moreover, crystallographic analyses have revealed that the phenyl moiety of the N-phenylpropyl substituent is accommodated in a previously unidentified subpocket within the active site of PfDXR. In this study, the crystal structures of PfDXR in complex with a series of reverse N-phenylalkyl derivatives of fosmidomycin were determined to ascertain whether the high inhibitory activities of the derivatives are consistently attributable to the utilization of the subpocket of PfDXR. While all reverse fosmidomycin derivatives with an N-substituted phenylalkyl group exhibit potent inhibitory activity against PfDXR, the present crystal structure analyses revealed that their binding modes to the PfDXR are not uniform. In these compounds, the nanomolar inhibitory activities appear to be driven by binding modes distinct from that observed for the inhibitor containing the N-phenylpropyl group. The structural information obtained in this study will provide a basis for further design of fosmidomycin derivatives.
Collapse
Affiliation(s)
- Sana Takada
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641, Japan (S.-i.O.)
| | - Mona A. Abdullaziz
- Institute of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
- National Research Centre (NRC), 33 El Buhouth St., Ad Doqi, Dokki, Cairo 12622, Egypt
| | - Stefan Höfmann
- Institute of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Talea Knak
- Institute of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Shin-ichiro Ozawa
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641, Japan (S.-i.O.)
| | - Yasumitsu Sakamoto
- School of Pharmacy, Iwate Medical University, Yahaba, Morioka 028-3694, Japan;
| | - Thomas Kurz
- Institute of Pharmaceutical and Medicinal Chemistry, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Nobutada Tanaka
- School of Pharmacy, Kitasato University, Minato-ku, Tokyo 108-8641, Japan (S.-i.O.)
| |
Collapse
|
6
|
Bizzarri L, Steinbrunn D, Quennesson T, Lacour A, Bianchino GI, Bravo P, Chaignon P, Lohse J, Mäser P, Seemann M, Calenbergh SV, Hirsch AKH, Hahne H. Studying Target-Engagement of Anti-Infectives by Solvent-Induced Protein Precipitation and Quantitative Mass Spectrometry. ACS Infect Dis 2024; 10:4087-4102. [PMID: 39566904 DOI: 10.1021/acsinfecdis.4c00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Antimicrobial resistance (AMR) poses a serious threat to global health. The rapid emergence of resistance contrasts with the slow pace of antimicrobial development, emphasizing the urgent need for innovative drug discovery approaches. This study addresses a critical bottleneck in early drug development by introducing integral solvent-induced protein precipitation (iSPP) to rapidly assess the target-engagement of lead compounds in extracts of pathogenic microorganisms under close-to-physiological conditions. iSPP measures the change in protein stability against solvent-induced precipitation in the presence of ligands. The iSPP method for bacteria builds upon established SPP procedures and features optimized denaturation gradients and minimized sample input amounts. The effectiveness of the iSPP workflow was initially demonstrated through a multidrug target-engagement study. Using quantitative mass spectrometry (LC-MS/MS), we successfully identified known drug targets of seven different antibiotics in cell extracts of four AMR-related pathogens: the three Gram-negative bacteria Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and the Gram-positive bacterium Staphylococcus aureus. The iSPP method was ultimately applied to demonstrate target-engagement of compounds derived from target-based drug discovery. We employed five small molecules targeting three enzymes in the 2-C-methyl-d-erythritol 4-phosphate (MEP) pathway─a promising focus for anti-infective drug development. The study showcases iSPP adaptability and efficiency in identifying anti-infective drug targets, advancing early-stage drug discovery against AMR.
Collapse
Affiliation(s)
- Lorenzo Bizzarri
- OmicScouts GmbH, Lise-Meitner-Straße 30, Freising D-85354, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
| | - Dominik Steinbrunn
- OmicScouts GmbH, Lise-Meitner-Straße 30, Freising D-85354, Germany
- TUM School of Natural Sciences, Department of Bioscience, Technical University of Munich, Center for Functional Protein Assemblies (CPA), Garching bei München D-85748, Germany
| | - Thibaut Quennesson
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, Gent B-9000, Belgium
| | - Antoine Lacour
- Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
- Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
| | - Gabriella Ines Bianchino
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg, UMR 7177, Université de Strasbourg/CNRS, 4, Rue Blaise Pascal, Strasbourg F-67070, France
| | - Patricia Bravo
- Swiss Tropical and Public Health Institute (Swiss TPH), Kreuzstrasse 2, Allschwil CH-4123, Switzerland
- University of Basel, Petersgraben 1, Basel CH-4001, Switzerland
| | - Philippe Chaignon
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg, UMR 7177, Université de Strasbourg/CNRS, 4, Rue Blaise Pascal, Strasbourg F-67070, France
| | - Jonas Lohse
- OmicScouts GmbH, Lise-Meitner-Straße 30, Freising D-85354, Germany
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute (Swiss TPH), Kreuzstrasse 2, Allschwil CH-4123, Switzerland
- University of Basel, Petersgraben 1, Basel CH-4001, Switzerland
| | - Myriam Seemann
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg, UMR 7177, Université de Strasbourg/CNRS, 4, Rue Blaise Pascal, Strasbourg F-67070, France
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, Gent B-9000, Belgium
| | - Anna K H Hirsch
- Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
- Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
| | - Hannes Hahne
- OmicScouts GmbH, Lise-Meitner-Straße 30, Freising D-85354, Germany
| |
Collapse
|
7
|
Willocx D, Bizzarri L, Alhayek A, Kannan D, Bravo P, Illarionov B, Rox K, Lohse J, Fischer M, Kany AM, Hahne H, Rottmann M, Witschel M, Odom John A, Hamed MM, Diamanti E, Hirsch AKH. Targeting Plasmodium falciparum IspD in the Methyl-d-erythritol Phosphate Pathway: Urea-Based Compounds with Nanomolar Potency on Target and Low-Micromolar Whole-Cell Activity. J Med Chem 2024; 67:17070-17086. [PMID: 39303294 PMCID: PMC11472328 DOI: 10.1021/acs.jmedchem.4c00212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/26/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
The methyl-d-erythritol phosphate (MEP) pathway has emerged as an interesting target in the fight against antimicrobial resistance. The pathway is essential in many human pathogens, including Plasmodium falciparum (Pf), but is absent in human cells. In the present study, we report on the discovery of a new chemical class targeting IspD, the third enzyme in the pathway. Exploration of the structure-activity relationship yielded inhibitors with potency in the low-nanomolar range. Moreover, we investigated the whole-cell activity, mode of inhibition, metabolic, and plasma stability of this compound class, and conducted in vivo pharmacokinetic profiling on selected compounds. Lastly, we disclosed a new mass spectrometry (MS)-based enzymatic assay for direct IspD activity determination, circumventing the need for auxiliary enzymes. In summary, we have identified a readily synthesizable compound class, demonstrating excellent activity and a promising profile, positioning it as a valuable tool compound for advancing research on IspD.
Collapse
Affiliation(s)
- Daan Willocx
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Department
of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Lorenzo Bizzarri
- Department
of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- OmicScouts
GmbH, Lise-Meitner-Straße
30, 85354 Freising, Germany
| | - Alaa Alhayek
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Deepika Kannan
- Department
of Pediatrics, Children’s Hospital
of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Patricia Bravo
- Swiss
Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Universität
Basel, Petersplatz 1, 4003 Basel, Switzerland
| | - Boris Illarionov
- Hamburg
School of Food Science, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany
| | - Katharina Rox
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Jonas Lohse
- OmicScouts
GmbH, Lise-Meitner-Straße
30, 85354 Freising, Germany
| | - Markus Fischer
- Hamburg
School of Food Science, University of Hamburg, Grindelallee 117, 20146 Hamburg, Germany
| | - Andreas M. Kany
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Hannes Hahne
- OmicScouts
GmbH, Lise-Meitner-Straße
30, 85354 Freising, Germany
| | - Matthias Rottmann
- Swiss
Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Universität
Basel, Petersplatz 1, 4003 Basel, Switzerland
| | | | - Audrey Odom John
- Department
of Pediatrics, Children’s Hospital
of Philadelphia, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Mostafa M. Hamed
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Eleonora Diamanti
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Anna K. H. Hirsch
- Helmholtz
Institute for Pharmaceutical Research (HIPS)-Helmholtz Centre for
Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Department
of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
8
|
Mazzone F, Hoeppner A, Reiners J, Gertzen CG, Applegate V, Abdullaziz MA, Gottstein J, Degrandi D, Wesemann M, Kurz T, Smits SH, Pfeffer K. 1-Deoxy-d-xylulose 5-phosphate reductoisomerase as target for anti Toxoplasma gondii agents: crystal structure, biochemical characterization and biological evaluation of inhibitors. Biochem J 2024; 481:1075-1096. [PMID: 39105673 PMCID: PMC11346426 DOI: 10.1042/bcj20240110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/27/2024] [Accepted: 08/06/2024] [Indexed: 08/07/2024]
Abstract
Toxoplasma gondii is a widely distributed apicomplexan parasite causing toxoplasmosis, a critical health issue for immunocompromised individuals and for congenitally infected foetuses. Current treatment options are limited in number and associated with severe side effects. Thus, novel anti-toxoplasma agents need to be identified and developed. 1-Deoxy-d-xylulose 5-phosphate reductoisomerase (DXR) is considered the rate-limiting enzyme in the non-mevalonate pathway for the biosynthesis of the isoprenoid precursors isopentenyl pyrophosphate and dimethylallyl pyrophosphate in the parasite, and has been previously investigated for its key role as a novel drug target in some species, encompassing Plasmodia, Mycobacteria and Escherichia coli. In this study, we present the first crystal structure of T. gondii DXR (TgDXR) in a tertiary complex with the inhibitor fosmidomycin and the cofactor NADPH in dimeric conformation at 2.5 Å resolution revealing the inhibitor binding mode. In addition, we biologically characterize reverse α-phenyl-β-thia and β-oxa fosmidomycin analogues and show that some derivatives are strong inhibitors of TgDXR which also, in contrast with fosmidomycin, inhibit the growth of T. gondii in vitro. Here, ((3,4-dichlorophenyl)((2-(hydroxy(methyl)amino)-2-oxoethyl)thio)methyl)phosphonic acid was identified as the most potent anti T. gondii compound. These findings will enable the future design and development of more potent anti-toxoplasma DXR inhibitors.
Collapse
Affiliation(s)
- Flaminia Mazzone
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University, Düsseldorf, Germany
- University Hospital Düsseldorf, Düsseldorf, Germany
| | - Astrid Hoeppner
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
| | - Jens Reiners
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
| | - Christoph G.W. Gertzen
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Violetta Applegate
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
| | - Mona A. Abdullaziz
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Düsseldorf, Germany
- National Research Centre (NRC), Dokki, Cairo, Egypt
| | - Julia Gottstein
- Institute of Biochemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Degrandi
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University, Düsseldorf, Germany
- University Hospital Düsseldorf, Düsseldorf, Germany
| | - Martina Wesemann
- Institute of Biochemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Thomas Kurz
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Sander H.J. Smits
- Center for Structural Studies, Heinrich Heine University, Düsseldorf, Germany
- Institute of Biochemistry, Heinrich Heine University, Düsseldorf, Germany
| | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University, Düsseldorf, Germany
- University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
9
|
Cui JJ, Zhang Y, Ju KS. Phosphonoalamides Reveal the Biosynthetic Origin of Phosphonoalanine Natural Products and a Convergent Pathway for Their Diversification. Angew Chem Int Ed Engl 2024; 63:e202405052. [PMID: 38780891 PMCID: PMC11867202 DOI: 10.1002/anie.202405052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 05/25/2024]
Abstract
Phosphonate natural products, with their potent inhibitory activity, have found widespread use across multiple industries. Their success has inspired development of genome mining approaches that continue to reveal previously unknown bioactive scaffolds and biosynthetic insights. However, a greater understanding of phosphonate metabolism is required to enable prediction of compounds and their bioactivities from sequence information alone. Here, we expand our knowledge of this natural product class by reporting the complete biosynthesis of the phosphonoalamides, antimicrobial tripeptides with a conserved N-terminal l-phosphonoalanine (PnAla) residue produced by Streptomyces. The phosphonoalamides result from the convergence of PnAla biosynthesis and peptide ligation pathways. We elucidate the biochemistry underlying the transamination of phosphonopyruvate to PnAla, a new early branchpoint in phosphonate biosynthesis catalyzed by an aminotransferase with evolved specificity for phosphonate metabolism. Peptide formation is catalyzed by two ATP-grasp ligases, the first of which produces dipeptides, and a second which ligates dipeptides to PnAla to produce phosphonoalamides. Substrate specificity profiling revealed a dramatic expansion of dipeptide and tripeptide products, while finding PnaC to be the most promiscuous dipeptide ligase reported thus far. Our findings highlight previously unknown transformations in natural product biosynthesis, promising enzyme biocatalysts, and unveil insights into the diversity of phosphonopeptide natural products.
Collapse
Affiliation(s)
- Jerry J. Cui
- Department of Microbiology, The Ohio State University, 318W. 12th Ave, Columbus, OH-43210 (USA)
| | - Yeying Zhang
- Department of Microbiology, The Ohio State University, 318W. 12th Ave, Columbus, OH-43210 (USA)
| | - Kou-San Ju
- Department of Microbiology, The Ohio State University, 318W. 12th Ave, Columbus, OH-43210 (USA)
- Division of Medicinal Chemistry and Pharmacognosy, Center for Applied Plant Sciences, Infectious Disease Institute, The Ohio State University, 318W. 12th Ave, Columbus, OH-43210 (USA)
| |
Collapse
|
10
|
Cui J, Ju KS. Biosynthesis of Bacillus Phosphonoalamides Reveals Highly Specific Amino Acid Ligation. ACS Chem Biol 2024; 19:1506-1514. [PMID: 38885091 PMCID: PMC11259534 DOI: 10.1021/acschembio.4c00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Phosphonate natural products have a history of commercial success across numerous industries due to their potent inhibition of metabolic processes. Over the past decade, genome mining approaches have successfully led to the discovery of numerous bioactive phosphonates. However, continued success is dependent upon a greater understanding of phosphonate metabolism, which will enable the prioritization and prediction of biosynthetic gene clusters for targeted isolation. Here, we report the complete biosynthetic pathway for phosphonoalamides E and F, antimicrobial phosphonopeptides with a conserved C-terminal l-phosphonoalanine (PnAla) residue. These peptides, produced by Bacillus, are the direct result of PnAla biosynthesis and serial ligation by two ATP-grasp ligases. A critical step of this pathway was the reversible transamination of phosphonopyruvate to PnAla by a dedicated transaminase with preference for the forward reaction. The dipeptide ligase PnfA was shown to ligate alanine to PnAla to afford phosphonoalamide E, which was subsequently ligated to alanine by PnfB to form phosphonoalamide F. Specificity profiling of both ligases found each to be highly specific, although the limited acceptance of noncanonical substrates by PnfA allowed for in vitro formation of products incorporating alternative pharmacophores. Our findings further establish the transaminative branch of phosphonate metabolism, unveil insights into the specificity of ATP-grasp ligation, and highlight the biocatalytic potential of biosynthetic enzymes.
Collapse
Affiliation(s)
- Jerry Cui
- Department of Microbiology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Kou-San Ju
- Department of Microbiology, The Ohio State University, Columbus, Ohio 43210, United States
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Applied Plant Sciences, The Ohio State University, Columbus, Ohio 43210, United States
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
11
|
Bague D, Wang R, Hodge D, Mikati MO, Roma JS, Boshoff HI, Dailey AL, Girma M, Couch RD, Odom John AR, Dowd CS. Inhibition of DXR in the MEP pathway with lipophilic N-alkoxyaryl FR900098 analogs. RSC Med Chem 2024; 15:2422-2439. [PMID: 39026652 PMCID: PMC11253873 DOI: 10.1039/d3md00642e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/22/2024] [Indexed: 07/20/2024] Open
Abstract
In Mycobacterium tuberculosis (Mtb) and Plasmodium falciparum (Pf), the methylerythritol phosphate (MEP) pathway is responsible for isoprene synthesis. This pathway and its products are vital to bacterial/parasitic metabolism and survival, and represent an attractive set of drug targets due to their essentiality in these pathogens but absence in humans. The second step in the MEP pathway is the conversion of 1-deoxy-d-xylulose-5-phosphate (DXP) to MEP and is catalyzed by 1-deoxy-d-xylulose-5-phosphate reductoisomerase (DXR). Natural products fosmidomycin and FR900098 inhibit DXR, but are too polar to reach the desired target inside some cells, such as Mtb. Synthesized FR900098 analogs with lipophilic substitution in the position α to the phosphorous atom showed promise, resulting in increased activity against Mtb and Pf. Here, an α substitution, consisting of a 3,4-dichlorophenyl substituent, in combination with various O-linked alkylaryl substituents on the hydroxamate moiety is utilized in the synthesis of a novel series of FR900098 analogs. The purpose of the O-linked alkylaryl substituents is to further enhance DXR inhibition by extending the structure into the adjacent NADPH binding pocket, blocking the binding of both DXP and NADPH. Of the initial O-linked alkylaryl substituted analogs, compound 6e showed most potent activity against Pf parasites at 3.60 μM. Additional compounds varying the phenyl ring of 6e were synthesized. The most potent phosphonic acids, 6l and 6n, display nM activity against PfDXR and low μM activity against Pf parasites. Prodrugs of these compounds were less effective against Pf parasites but showed modest activity against Mtb cells. Data from this series of compounds suggests that this combination of substituents can be advantageous in designing a new generation of antimicrobials.
Collapse
Affiliation(s)
- Darean Bague
- Department of Chemistry, George Washington University Washington D.C. 20052 USA
| | - Ruiqin Wang
- Department of Chemistry, George Washington University Washington D.C. 20052 USA
| | - Dana Hodge
- Division of Infectious Diseases, Children's Hospital of Philadelphia Philadelphia PA 19104 USA
| | - Marwa O Mikati
- Department of Molecular Microbiology, Washington University School of Medicine St. Louis MO 63110 USA
| | - Jose S Roma
- Tuberculosis Research Section, LCIM, NIAID/NIH Bethesda MD 20892 USA
| | - Helena I Boshoff
- Tuberculosis Research Section, LCIM, NIAID/NIH Bethesda MD 20892 USA
| | - Allyson L Dailey
- Department of Chemistry and Biochemistry, George Mason University Fairfax VA 22030 USA
| | - Misgina Girma
- Department of Chemistry and Biochemistry, George Mason University Fairfax VA 22030 USA
| | - Robin D Couch
- Department of Chemistry and Biochemistry, George Mason University Fairfax VA 22030 USA
| | - Audrey R Odom John
- Division of Infectious Diseases, Children's Hospital of Philadelphia Philadelphia PA 19104 USA
- Department of Molecular Microbiology, Washington University School of Medicine St. Louis MO 63110 USA
| | - Cynthia S Dowd
- Department of Chemistry, George Washington University Washington D.C. 20052 USA
| |
Collapse
|
12
|
Schwarzer E, Skorokhod O. Post-Translational Modifications of Proteins of Malaria Parasites during the Life Cycle. Int J Mol Sci 2024; 25:6145. [PMID: 38892332 PMCID: PMC11173270 DOI: 10.3390/ijms25116145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Post-translational modifications (PTMs) are essential for regulating protein functions, influencing various fundamental processes in eukaryotes. These include, but are not limited to, cell signaling, protein trafficking, the epigenetic control of gene expression, and control of the cell cycle, as well as cell proliferation, differentiation, and interactions between cells. In this review, we discuss protein PTMs that play a key role in the malaria parasite biology and its pathogenesis. Phosphorylation, acetylation, methylation, lipidation and lipoxidation, glycosylation, ubiquitination and sumoylation, nitrosylation and glutathionylation, all of which occur in malarial parasites, are reviewed. We provide information regarding the biological significance of these modifications along all phases of the complex life cycle of Plasmodium spp. Importantly, not only the parasite, but also the host and vector protein PTMs are often crucial for parasite growth and development. In addition to metabolic regulations, protein PTMs can result in epitopes that are able to elicit both innate and adaptive immune responses of the host or vector. We discuss some existing and prospective results from antimalarial drug discovery trials that target various PTM-related processes in the parasite or host.
Collapse
Affiliation(s)
- Evelin Schwarzer
- Department of Oncology, University of Turin, Via Santena 5 bis, 10126 Turin, Italy;
| | - Oleksii Skorokhod
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina, 13, 10123 Turin, Italy
| |
Collapse
|
13
|
Chen X, Zhao H, Wang C, Hamed M, Shang Q, Yang Y, Diao X, Sun X, Hu W, Jiang X, Zhang Y, Hirsch AKH, Wu D, Zhuang J. Two natural compounds as potential inhibitors against the Helicobacter pylori and Acinetobacter baumannii IspD enzymes. Int J Antimicrob Agents 2024; 63:107160. [PMID: 38537721 DOI: 10.1016/j.ijantimicag.2024.107160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/27/2024] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
In a vast majority of bacteria, protozoa and plants, the methylerythritol phosphate (MEP) pathway is utilized for the synthesis of isopentenyl diphosphate (IDP) and dimethylallyl diphosphate (DMADP), which are precursors for isoprenoids. Isoprenoids, such as cholesterol and coenzyme Q, play a variety of crucial roles in physiological activities, including cell-membrane formation, protein degradation, cell apoptosis, and transcription regulation. In contrast, humans employ the mevalonate (MVA) pathway for the production of IDP and DMADP, rendering proteins in the MEP pathway appealing targets for antimicrobial agents. This pathway consists of seven consecutive enzymatic reactions, of which 4-diphosphocytidyl-2C-methyl-D-erythritol synthase (IspD) and 2C-methyl-D-erythritol 2,4-cyclodiphosphate synthase (IspF) catalyze the third and fifth steps, respectively. In this study, we characterized the enzymatic activities and protein structures of Helicobacter pylori IspDF and Acinetobacter baumannii IspD. Then, using the direct interaction-based thermal shift assay, we conducted a compound screening of an approved drug library and identified 27 hit compounds potentially binding to AbIspD. Among them, two natural products, rosmarinic acid and tanshinone IIA sodium sulfonate, exhibited inhibitory activities against HpIspDF and AbIspD, by competing with one of the substrates, MEP. Moreover, tanshinone IIA sodium sulfonate also demonstrated certain antibacterial effects against H. pylori. In summary, we identified two IspD inhibitors from approved ingredients, broadening the scope for antibiotic discovery targeting the MEP pathway.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Huilin Zhao
- Department of Pathogenic Biology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Chuandong Wang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Mostafa Hamed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbrücken, Germany
| | - Qinghong Shang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Yating Yang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiaotong Diao
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiangnan Sun
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Wei Hu
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xukai Jiang
- National Glycoengineering Research Center, Shandong University, Qingdao, China
| | - Youming Zhang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Dalei Wu
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China.
| | - Jingjing Zhuang
- Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China; Marine College, Shandong University, Weihai, China.
| |
Collapse
|
14
|
Wu X, Yang Z, Song C, Bu M, Li W, Duan J, Yang GF, Zhang A. Hydroxamate-Containing Bisphosphonates as Fosmidomycin Analogues: Design, Synthesis, and Proherbicide Activity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7684-7693. [PMID: 38532701 DOI: 10.1021/acs.jafc.3c07872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Fosmidomycin (FOS) is a natural product inhibiting the DXR enzyme in the MEP pathway and has stimulated interest for finding more suitable FOS analogues. Herein, two series of FOS analogue hydroxamate-containing bisphosphonates as proherbicides were designed, with bisphosphonate replacing the phosphonic unit in FOS while retaining the hydroxamate (BPF series) or replacing it with retro-hydroxamate (BPRF series). The BPF series were synthesized through a three-step reaction sequence including Michael addition of vinylidenebisphosphonate, N-acylation, and deprotection, and the BPRF series were synthesized with a retro-Claisen condensation incorporated into the reaction sequence. Evaluation on model plants demonstrated several compounds having considerable herbicidal activities, and in particular, compound 8m exhibited multifold activity enhancement as compared to the control FOS. The proherbicide properties were comparatively validated. Furthermore, DXR enzyme assay, dimethylallyl pyrophosphate rescue, and molecular docking verified 8m to be a promising proherbicide candidate targeting the DXR enzyme. In addition, 8m also displayed good antimalarial activities.
Collapse
Affiliation(s)
- Xin Wu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Zili Yang
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Chunlin Song
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Mengwei Bu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Weiguo Li
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Jiang Duan
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Guang-Fu Yang
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Aidong Zhang
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China
| |
Collapse
|
15
|
Wu X, Bu M, Yang Z, Ping H, Song C, Duan J, Zhang A. Design and synthesis of fosmidomycin analogs containing aza-linkers and their biological activity evaluation. PEST MANAGEMENT SCIENCE 2024; 80:846-856. [PMID: 37794283 DOI: 10.1002/ps.7810] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND The enzymes involved in the 2-C-methyl-d-erythritol 4-phosphate (MEP) pathway are attractive targets of a new mode of action for developing anti-infective drugs and herbicides, and inhibitors against 1-deoxy-d-xylulose 5-phosphate reductoisomerase (IspC), the second key enzyme in the pathway, have been intensively investigated; however, few works are reported regarding IspC inhibitors designed for new herbicide discovery. RESULTS A series of fosmidomycin (FOS) analogs were designed with nitrogen-containing linkers replacing the trimethylene linker between the two active substructures of FOS, phosphonic acid and hydroxamic acid. Synthesis followed a facile three-step route of sequential aza-Michael addition of α-amino acids to dibenzyl vinylphosphonate, amidation of the amino acid carboxyl with O-benzyl hydroxylamine, and simultaneous removal of the benzyl protective groups. Biological activity evaluation of IspC and model plants revealed that some compounds had moderate enzyme and model plant growth inhibition effects. In particular, compound 10g, which has a N-(4-fluorophenylethyl) nitrogen-containing linker, exhibited the best plant inhibition activities, superior to the control FOS against the model plants Arabidopsis thaliana, Brassica napus L., Amaranthus retroflexus and Echinochloa crus-galli. A dimethylallyl pyrophosphate rescue assay on A. thaliana confirmed that both 10g and FOS exert their herbicidal activity by blocking the MEP pathway. This result consistent with molecular docking, which confirmed 10g and FOS binding to the IspC active site in a similar way. CONCLUSION Compound 10g has excellent herbicidal activity and represents the first herbicide lead structure of a new mode of action that targets IspC enzyme in the MEP pathway. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xin Wu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Mengwei Bu
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Zili Yang
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Hongrui Ping
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Chunlin Song
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Jiang Duan
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| | - Aidong Zhang
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan, China
| |
Collapse
|
16
|
Hamid A, Mäser P, Mahmoud AB. Drug Repurposing in the Chemotherapy of Infectious Diseases. Molecules 2024; 29:635. [PMID: 38338378 PMCID: PMC10856722 DOI: 10.3390/molecules29030635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Repurposing is a universal mechanism for innovation, from the evolution of feathers to the invention of Velcro tape. Repurposing is particularly attractive for drug development, given that it costs more than a billion dollars and takes longer than ten years to make a new drug from scratch. The COVID-19 pandemic has triggered a large number of drug repurposing activities. At the same time, it has highlighted potential pitfalls, in particular when concessions are made to the target product profile. Here, we discuss the pros and cons of drug repurposing for infectious diseases and analyze different ways of repurposing. We distinguish between opportunistic and rational approaches, i.e., just saving time and money by screening compounds that are already approved versus repurposing based on a particular target that is common to different pathogens. The latter can be further distinguished into divergent and convergent: points of attack that are divergent share common ancestry (e.g., prokaryotic targets in the apicoplast of malaria parasites), whereas those that are convergent arise from a shared lifestyle (e.g., the susceptibility of bacteria, parasites, and tumor cells to antifolates due to their high rate of DNA synthesis). We illustrate how such different scenarios can be capitalized on by using examples of drugs that have been repurposed to, from, or within the field of anti-infective chemotherapy.
Collapse
Affiliation(s)
- Amal Hamid
- Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan;
| | - Pascal Mäser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, 4123 Basel, Switzerland
- Faculty of Science, University of Basel, 4001 Basel, Switzerland
| | - Abdelhalim Babiker Mahmoud
- Faculty of Pharmacy, University of Khartoum, Khartoum 11111, Sudan;
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland, 66123 Saarbruecken, Germany
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| |
Collapse
|
17
|
Voráčová M, Zore M, Yli-Kauhaluoma J, Kiuru P. Harvesting phosphorus-containing moieties for their antibacterial effects. Bioorg Med Chem 2023; 96:117512. [PMID: 37939493 DOI: 10.1016/j.bmc.2023.117512] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023]
Abstract
Clinically manifested resistance of bacteria to antibiotics has emerged as a global threat to society and there is an urgent need for the development of novel classes of antibacterial agents. Recently, the use of phosphorus in antibacterial agents has been explored in quite an unprecedent manner. In this comprehensive review, we summarize the use of phosphorus-containing moieties (phosphonates, phosphonamidates, phosphonopeptides, phosphates, phosphoramidates, phosphinates, phosphine oxides, and phosphoniums) in compounds with antibacterial effect, including their use as β-lactamase inhibitors and antibacterial disinfectants. We show that phosphorus-containing moieties can serve as novel pharmacophores, bioisosteres, and prodrugs to modify pharmacodynamic and pharmacokinetic properties. We further discuss the mechanisms of action, biological activities, clinical use and highlight possible future prospects.
Collapse
Affiliation(s)
- Manuela Voráčová
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Matej Zore
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Paula Kiuru
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
18
|
Theuretzbacher U, Blasco B, Duffey M, Piddock LJV. Unrealized targets in the discovery of antibiotics for Gram-negative bacterial infections. Nat Rev Drug Discov 2023; 22:957-975. [PMID: 37833553 DOI: 10.1038/s41573-023-00791-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 10/15/2023]
Abstract
Advances in areas that include genomics, systems biology, protein structure determination and artificial intelligence provide new opportunities for target-based antibacterial drug discovery. The selection of a 'good' new target for direct-acting antibacterial compounds is the first decision, for which multiple criteria must be explored, integrated and re-evaluated as drug discovery programmes progress. Criteria include essentiality of the target for bacterial survival, its conservation across different strains of the same species, bacterial species and growth conditions (which determines the spectrum of activity of a potential antibiotic) and the level of homology with human genes (which influences the potential for selective inhibition). Additionally, a bacterial target should have the potential to bind to drug-like molecules, and its subcellular location will govern the need for inhibitors to penetrate one or two bacterial membranes, which is a key challenge in targeting Gram-negative bacteria. The risk of the emergence of target-based drug resistance for drugs with single targets also requires consideration. This Review describes promising but as-yet-unrealized targets for antibacterial drugs against Gram-negative bacteria and examples of cognate inhibitors, and highlights lessons learned from past drug discovery programmes.
Collapse
Affiliation(s)
| | - Benjamin Blasco
- Global Antibiotic Research and Development Partnership (GARDP), Geneva, Switzerland
| | - Maëlle Duffey
- Global Antibiotic Research and Development Partnership (GARDP), Geneva, Switzerland
| | - Laura J V Piddock
- Global Antibiotic Research and Development Partnership (GARDP), Geneva, Switzerland.
| |
Collapse
|
19
|
Wu X, Yang Z, Bu M, Duan J, Zhang A. Design, Synthesis and Bioactivity Evaluation of Heterocycle-Containing Mono- and Bisphosphonic Acid Compounds. Molecules 2023; 28:7509. [PMID: 38005231 PMCID: PMC10673511 DOI: 10.3390/molecules28227509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Fosmidomycin (FOS) is a naturally occurring compound active against the 1-deoxy-D-xylulose 5-phosphate reductoisomerase (DXR) enzyme in the 2-C-methyl-D-erythritol 4-phosphate (MEP) pathway, and using it as a template for lead structure design is an effective strategy to develop new active compounds. In this work, by replacing the hydroxamate unit of FOS with pyrazole, isoxazole and the related heterocycles that also have metal ion binding affinity, while retaining the monophosphonic acid in FOS or replacing it with a bisphosphonic acid group, heterocycle-containing mono- and bisphosphonic acid compounds as FOS analogs were designed. The key steps involved in the facile synthesis of these FOS analogs included the Michael addition of diethyl vinylphosphonate or tetraethyl vinylidenebisphosphonate to β-dicarbonyl compounds and the subsequent cyclic condensation with hydrazine or hydroxylamine. Two additional isoxazolinone-bearing FOS analogs were synthesized via the Michaelis-Becker reaction with diethyl phosphite as a key step. The bioactivity evaluation on model plants demonstrated that several compounds have better herbicidal activities compared to FOS, with the most active compound showing a 3.7-fold inhibitory activity on Arabidopsis thaliana, while on the roots and stalks of Brassica napus L. and Echinochloa crus-galli in a pre-emergence inhibitory activity test, the activities of this compound were found to be 3.2- and 14.3-fold and 5.4- and 9.4-fold, respectively, and in a post-emergency activity test on Amaranthus retroflexus and Echinochloa crus-galli, 2.2- and 2.0-fold inhibition activities were displayed. Despite the significant herbicidal activity, this compound exhibited a DXR inhibitory activity lower than that of FOS but comparable to that of other non-hydroxamate DXR inhibitors, and the dimethylallyl pyrophosphate rescue assay gave no statistical significance, suggesting that a different target might be involved in the inhibiting process. This work demonstrates that using bioisosteric replacement can be considered as a valuable strategy to discover new FOS analogs that may have high herbicidal activities.
Collapse
Affiliation(s)
| | | | | | - Jiang Duan
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China; (X.W.); (Z.Y.); (M.B.)
| | - Aidong Zhang
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China; (X.W.); (Z.Y.); (M.B.)
| |
Collapse
|